You are on page 1of 13

Expert Review of Cardiovascular Therapy

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ierk20

Pathophysiology and mechanisms of Acute


Coronary Syndromes: atherothrombosis, immune-
inflammation, and beyond

Giovanni Cimmino, Luigi Di Serafino & Plinio Cirillo

To cite this article: Giovanni Cimmino, Luigi Di Serafino & Plinio Cirillo (2022)
Pathophysiology and mechanisms of Acute Coronary Syndromes: atherothrombosis, immune-
inflammation, and beyond, Expert Review of Cardiovascular Therapy, 20:5, 351-362, DOI:
10.1080/14779072.2022.2074836

To link to this article: https://doi.org/10.1080/14779072.2022.2074836

Published online: 17 May 2022.

Submit your article to this journal

Article views: 189

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierk20
EXPERT REVIEW OF CARDIOVASCULAR THERAPY
2022, VOL. 20, NO. 5, 351–362
https://doi.org/10.1080/14779072.2022.2074836

REVIEW

Pathophysiology and mechanisms of Acute Coronary Syndromes:


atherothrombosis, immune-inflammation, and beyond
Giovanni Cimminoa, Luigi Di Serafinob and Plinio Cirillo b

a
Department of Translational Medical Sciences, Section of Cardiology, University of Campania “Luigi Vanvitelli”, Naples, Italy; bDepartment of
Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy

ABSTRACT ARTICLE HISTORY


Introduction: The pathophysiology of atherosclerosis and its acute complications, such as the Acute Received 26 January 2022
Coronary Syndromes (ACS), is continuously under investigation. Immunity and inflammation seem to Accepted 4 May 2022
play a pivotal role in promoting formation and grow of atherosclerotic plaques. At the same time, plaque KEYWORDS
rupture followed by both platelets’ activation and coagulation cascade induction lead to intracoronary Atherosclerosis; Immune-
thrombus formation. Although these phenomena might be considered responsible of about 90% of ACS, in inflammation; MINOCA;
up to 5–10% of acute syndromes, a non-obstructive coronary artery disease (MINOCA) might be documen­ thrombosis
ted. This paper gives an overview on atherothrombosis and immuno-inflammation processes involved in
ACS pathophysiology, also emphasizing the pathological mechanisms potentially involved in MINOCA.
Areas covered: The relationship between immuno-inflammation and atherothrombosis is continuously
updated by recent findings. At the same time, pathophysiology of MINOCA still remains a partially
unexplored field, stimulating the research of potential links between these two aspects of ACS
pathophysiology.
Expert opinion: Pathophysiology of ACS has been extensively investigated; however, several gray areas
still remain. MINOCA represents one of these areas. At the same time, many aspects of immune-
inflammation processes are still unknown. Thus, research should be continued to shed a brighter light on
both these sides of “ACS” moon.

1. Introduction
atherothrombosis because coronary angiography does not
Atherosclerosis represents the anatomo-pathological substrate reveal a culprit lesion. In these cases, a functional alteration
of Acute Coronary Syndromes (ACS) in more than 90% of cases of coronary circulation involving either large epicardial coron­
[1–3]. A large mass of studies have extensively investigated ary arteries or the coronary microcirculation has been indi­
how atherosclerosis begins and develops, and it is now recog­ cated as responsible of the acute event [6]. Specifically,
nized that it might be considered a chronic immune- macrovascular and microvascular spasms have been postu­
inflammatory disease starting with deposition of lipids within lated to be responsible of ischemia with the clinical manifesta­
the intima of the vessels, involving blood cells belonging to tion of non-obstructive coronary artery diseases (CADs)
both innate and acquired immune system [1], and then, it (MINOCA) [6,7].
progresses with cellular and inflammatory response against Although mechanisms of atherothrombosis have been
undefined antigens [4] finally causing plaque grow and for­ extensively investigated and the biological pathways under­
mation of a “stable” stenosis of arterial vessels. When plaque lying the conversion from asymptomatic or stable chronic
becomes “unstable” as consequence of rupture, erosion, and/ angina to ACS have been widely studied, the complex patho­
or ulceration, activation of both coagulation cascade and pla­ physiological puzzle of this disease has still some gray areas.
telets causes intravascular thrombus formation and thrombo­ At the same time, the basic mechanisms by which cardiovas­
tic vessel occlusion. Therefore, the term atherothrombosis has cular risk factors might induce coronary reserve reduction
been proposed to better describe this process [5]. Clinical (because of non-angiographically significant atherosclerotic
manifestations of stable coronary atherosclerotic stenosis are stenosis or functional alteration of coronary circulation)
represented by effort angina or dyspnea associated with tran­ remain to be better elucidated.
sient ECG signs, which occur when coronary reserve is signifi­ In this article, we first provide an overview on the main
cantly reduced. On the contrary, rest angina and persistent mechanisms involved in pathophysiology of ACS, and then,
ECG modifications are signs of abrupt coronary blood flow we discuss the hypothetical pathological mechanisms to
reduction due to plaque rupture and coronary thrombosis explain how functional alterations of the coronary circulation
[6]. As reported in Table 1, in about 10% of patients with (at both macrovascular and microvascular level) might be
ACS, acute myocardial ischemia is not associated with responsible of the clinical manifestations of ACS.

CONTACT Plinio Cirillo pcirillo@unina.it Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy, Via S. Pansini 5,
Bilding 2- Naples, Italy
© 2022 Informa UK Limited, trading as Taylor & Francis Group
352 G. CIMMINO ET AL.

lesion formation [12]. At this stage, retention and modification


Article highlights of LDL particles (mainly the oxidized forms, oxLDL) and sub­
● Atherothrombosis plays a key role in the majority of ACS: plaque
sequent accumulation in the intima are the initial trigger of
rupture exposes intraplaque thrombogenic components to the flow­ the endothelial dysfunction [13]. High levels of LDL cholesterol
ing blood, such as TF, leading to coagulation cascade activation with seem to be the major determinant for this effect [14]. Most of
final intravascular thrombus formation.
● The immuno-inflammatory modulation of atherosclerosis with still
the LDL actions are mediated by apoprotein B (Apo B) that is
unknown “intraplaque antigen” activation of the immunocompetent its major protein component. This protein mediates the inter­
cells is now well documented and accepted. action with the endothelial cells surface via different matrix
● Platelet aggregation cannot be considered only the final event of
coagulation cascade. Activation of platelets occurs via a complex
proteins such as proteoglycan, collagen, and fibronectin [14–
process that involves modulation of the proteome, transcriptome, 16]. Within the intimal microenvironment, LDL may be mod­
and miRNOme. ified, mainly oxidized by lipoxygenase, myeloperoxidase, or
● A not negligible percentage of ACS is related to non-obstructive
CADs (MINOCA) where a macrovascular or microvascular disorder
reactive oxygen species released in loco, thus damaging the
may occur. endothelium. The final result of this interaction is the activa­
● Several gray areas remain to be explored: 1) the identification of tion of some transcription factors, such as NF-kappa B, leading
possible antigens responsible of activation of immuno-inflammation
cells; 2) the definition of platelet role beyond thrombosis; and 3) the
to a modulation of the gene expression profile and up-
improvement of our knowledge on the pathophysiology of MINOCA regulation of adhesion molecules (CAMs), mainly VCAM1,
to provide the patients with the best tailored therapy. and selectins. Binding of these proteins with other membrane
proteins on circulating immune cells will facilitate their adhe­
sion and migration leading to plaque grow [17,18]. Recent
studies have indicated that within the subendothelial space,
2. Atherothrombosis: a history that starts with LDL particles may aggregate by binding intimal proteoglycans.
endothelial dysfunction, continues with lipids These aggregates may infiltrate smooth muscle cells (SMCs)
deposition, and ends with thrombus formation via specific receptors belonging to the LDL receptor-related
2.1. Pathophysiology of plaque grow protein superfamily in LDL receptor independent manner [19].
Thus, non-only macrophages but also SMCs finally contribute
Endothelial dysfunction has been recognized as the initial step to lesion formation [19,20]. Moreover, SMCs may also partici­
of atherosclerosis [8]. In physiological condition, endothelium pate to the atherosclerotic plaque grow because by migrating
exerts several anti-atherothrombotic functions because it has from the media into the intima they accumulate at the site of
anti-inflammatory and anti-mitogenic properties, contractile lesion formation. Finally, these cells, beyond a continuous
activities, and regulatory effects of the hemostasis process proliferation lasting for years, are able to produce and release
[9]. The conventional risk factors such as hypercholesterole­ extracellular matrix degrading enzyme that may induce ather­
mia, hypertension, diabetes mellitus, and smoke are the main osclerotic plaque rupture or erosion [21].
determinants of endothelial dysfunction [10,11]. Once However, besides these mechanisms, plaque growth might
endothelial dysfunction occurs, atherosclerosis continues be due to cycles of repeated plaque ruptures and healing.
with lipid deposition within the arterial wall. This phenom­ Indeed, plaque rupture does not always lead to an acute
enon seems to be the consequence of an unbalance between fatal event since the majority of ruptures and/or erosions are
the “influx” of cholesterol (mainly related to low-density lipo­ asymptomatic. In fact, autoptic studies in patients who died of
proteins) and “efflux” of cholesterol (mainly high-density lipo­ noncardiovascular causes and studies with imaging of coron­
protein), thus leading to the primary step of atherosclerotic ary tree have shown that plaque rupture was found in a small

Table 1. Main causes of Acute Coronary Syndromes according to pathological substrate.


Pathological
Anatomical localization Causes substrate Diagnostic Tools
Plaque-related disease Epicardial compartment Atherothrombosis Atherosclerotic Invasive Coronary
Angiography
Epicardial and microvascular Plaque rupture/erosion and distal Atherosclerotic Invasive Coronary
compartment embolization Angiography
and Intracoronary Imaging
(IVUS or OCT)
Coronary microvascular Epicardial compartment Epicardial vasospasm Non- Invasive Coronary
dysfunction atherosclerotic Angiography
and Functional Assessment
(FFR/IMR/CFR/
Acetylcholine Test)
Microvascular compartment Microvascular spasm and/or Non- Invasive Coronary
structural remodeling atherosclerotic Angiography
and Functional Assessment
(FFR/IMR/CFR/
Acetylcholine Test)
CRF: Coronary Flow Reserve; FFR: Fractional Flow Reserve; IMR: Index of Microcirculatory Resistance; IVUS: Intravascular Ultrasound; OCT: Optical Coherence
Tomography
EXPERT REVIEW OF CARDIOVASCULAR THERAPY 353

number (less than 10%) of atherosclerotic lesions studied However, an increasing number of evidence, including several
[22,23]. In those lesions in which plaque rupture is not fol­ reports by our group, have focused on the role of inflamma­
lowed by an occlusive thrombosis [22], the thrombus may tion (local and systemic) as well as the on-site immune-
remain mural with incomplete lysis. The healing phase con­ competent cells activation (both natural and adaptive
tinues with thrombus reendothelialization followed by fibrous immunity) [3,28–30]. On this regard, it has been reported by
thrombus organization: this phenomenon finally causes expo­ several functional and morphological studies that complicated
nential plaque growth [24]. plaques are more inflamed and enriched of immune-
inflammatory cells than intact plaques [28,29] with increased
neovascularity and intraplaque hemorrhage [31,32].
2.2. The concept of vulnerability: a prone to rupture Inflammation may exert different actions: the release of col­
lesion lagen degrading enzymes, such as metalloproteinases, by
Atherosclerotic plaque grow is associated with remodeling of endothelial cells, SMCs, macrophages or even platelets, thus
vessel wall: the lesions expand outward preserving the caliber inducing fibrous cap rupture, and the death of collagen-
of the arterial lumen and then it begins to expand upon the synthesizing SMCs, thus contributing to loss of fibrous cap
arterial lumen, finally causing the formation of lesions that integrity [28,32,33].
might progressively became “flow-limiting.” This is the case In the last two decades, the concept of vulnerability has
of stable coronary plaques characterized by limited lipid accu­ been proposed to describe plaques at high risk of imminent
mulation and thicker fibrous caps, usually responsible of rupture and thrombosis [34]. Specifically, it has been postu­
symptoms of effort angina pectoris [25]. lated that plaque vulnerability might be considered as
Atherosclerotic plaque rupture is the most common trigger of a biomechanical phenomenon with a complex interplay
ACS (Figure 1, Panel A) [6,26]. Ruptured plaques often have large between different factors including applied plaque stresses,
lipid cores covered by a thin fibrous cap [15]. Disruption of the structural features, and biological processes.
atherosclerotic plaque causes exposure of thrombogenic sub­ The prototypical vulnerable plaque is usually small, with
stances, mainly tissue factor (TF) to the flowing blood [27]. TF thin-cap fibroatheromas. Large necrotic core, high macro­
represents the natural initiator of extrinsic coagulation pathway, phage infiltration/activity in the cap, expansive remodeling
and its exposure to other coagulation factors, mainly FVII and X, mitigating luminal obstruction (mild stenosis by angiography),
contained in the blood stream, induces the activation of coagu­ neovascularization, plaque hemorrhage, adventitial inflamma­
lation cascade and, as final event, platelet aggregation via several tion, and a spotty pattern of calcifications are common fea­
membrane receptors (Figure 1, Panel B), leading to the intracor­ tures [24,31,32,35].
onary thrombus formation (Figure 1, Panel A) [2,27]. In the last years, several epidemiological studies have
The mechanisms involved into the translation from a stable clearly indicated the close relationship between cold tempera­
to an unstable atherosclerotic plaque remains to be clarified. ture and acute coronary events [36–38]. Several hypothetical

Figure 1. Panel a: Atherosclerotic plaque formation is the final event of a complex network involved lipid deposition, endothelial dysfunction, white blood cells
recruitment, and activation. Intravascular thrombus formation occurs on a complicated atherosclerotic lesion with exposure of TF, coagulation cascade activation,
and platelet aggregation. Panel b: Platelets express different membrane receptors which bind several agonists able to induce a deep proteome modulation via
miRNAs activation and mRNAs alternatively splicing. Panel c: Within the atherosclerotic plaque, antigen-presenting cells may modulate the immune response by
interacting with naive T-cells, thus becoming effector T-cells. According to the stimuli, the interactions between various T-cell subsets may be generated (Th1, Th2,
Th17, Treg), thus defining the final fate of the lesion. Specifically, Th1 and Th17 seem to be associated with a progression/destabilization of atherosclerotic lesion.
Th2 and Treg are linked to regulatory/inhibitory effects.
354 G. CIMMINO ET AL.

mechanisms have been proposed to explain how cold tem­ chemokines, including MCP-1 [71], proliferation and differen­
perature might be involved in the pathophysiology of cardio­ tiation of SMCs, activation of monocytes, macrophages, and
vascular events. It has been suggested that catecholamines secretion of different inflammatory mediators [70,72], and
plasma levels increase after stimulation of skin cold receptors. generation of IL-6 and MMPs [72] that are closely related to
This, in turn, causes vasoconstriction with increased blood the fibrous cap rupture as indicated above. Noteworthy, the
pressure together with increased heart rate finally leading to CANTOS study has highlighted that anti-inflammatory therapy
plaque rupture and acute coronary events [39,40]. Moreover, it with canakinumab, a monoclonal antibody against the IL-1β
has been suggested that acute events might be due to pathway, could significantly reduce the rate of recurrent car­
enhanced thrombotic milieu for increased plasma concentra­ diovascular events [53], thus reinforcing the hypothesis that
tions of clotting factors and platelets because cold tempera­ this inflammatory molecule plays an active role in pathophy­
ture might significantly increase diuresis and, consequently, siology of acute coronary events.
blood viscosity [41]. Lastly, other studies have underlined that IL-6 is also
Finally, a minor cause of ACS may be coronary embolism involved in atherosclerosis. Specifically, it promotes the devel­
[42]. Three types of coronary embolisms are known: 1) direct, 2) opment and destabilization of atherosclerotic plaques [52]. It
paradoxical, and 3) iatrogenic. Direct coronary emboli com­ can be released by cardiomyocytes during myocardial ische­
monly originate mainly from the left atrial appendage in mia-reperfusion, inducing CAMs expression with final damage
patients with atrial fibrillation who are not treated with antic­ of the myocardium from the immune cells [73]. Moreover, IL-6
oagulants [43]. Again, another cause of direct coronary embo­ is elevated in animal model of cerebral ischemia and in the
lism might be a thrombus of the left ventricle specially in cerebrospinal fluid of patients with acute stroke [73].
patients with severe ventricular dysfunction or aneurysm
[44]. Finally, other sources might be the prosthetic aortic or
mitral valves [45]. The main cause of paradoxical coronary
3.2. Role of immunity
embolisms is patent foramen ovale [46]. Finally, iatrogenic
coronary embolisms might be secondary to coronary interven­ The concept that an active immune response occurs in ather­
tions [42]. osclerosis as well as in ischemic myocardium is now well
accepted [74]. Enrichment of immune-competent cells within
atherosclerotic lesions has been reported by our group [75]
3. Atherothrombosis: the role of and others [76,77], and contribution to plaque vulnerability is
immuno-inflammation also proposed [28,78]. Beyond the role of monocytes/macro­
phages since the initial stage of plaque formation in ingest­
3.1. Role of inflammation
ing lipids and orchestrating immunological response by
In the last two decades, several epidemiological studies have releasing inflammatory mediators [79], other white blood
clearly shown that some systemic inflammatory markers, such cells are involved in atherosclerosis, such as B and
as C-reactive protein (CRP) Interleukin-1 beta and Interleukin- T-lymphocytes [29,80]. Despite T-cells are a minority of the
6, might be useful prognostic markers to predict major car­ leukocytes enriching the plaques, several reports suggest
diovascular events in healthy subjects as well as in patients their key regulatory role within atherosclerotic lesion [81]
with established cardiovascular disease [47–53]. CRP was the by modulating the adaptive immune response and instruct­
first inflammatory marker that was correlated with cardiovas­ ing the more abundant monocytes/macrophages of the
cular diseases. This protein was measured within the coron­ innate immune response [28,82]. Moreover, atherosclerotic
ary circulation of ACS patients and in patients with lesions are also infiltrated by several antigen-presenting
cerebrovascular events [54–56]. However, more recent stu­ cells that in cooperation with soluble mediators released in
dies have clearly indicated that CRP might be no more con­ loco by activated macrophages may act simultaneously indu­
sidered as a simple marker but as an active partaker in ACS cing T-cells differentiation in different subsets [83]. Several
pathophysiology. Indeed, several in vitro and in vivo studies reports indicate that this polarization seems to define plaque
have shown that CRP exerts direct biological effects on destiny, since some T-cells subtypes exert more proinflamma­
endothelial cells: it promotes expression of adhesion mole­ tory effects (i.e. Th1 and Th17 cells), while other subtypes
cules [57–60] and TF [61], stimulates release of MCP-1 [62] may act as negative regulators of inflammation (Treg and Th2
and MMPs [63,64], secretion of other inflammatory cytokines cells) [28,84]. T-cell activation and polarization are crucial for
[65,66], increases iNOS and superoxide production, and plaque vulnerability [32,33,85]. It has been reported by our
decreases eNOS, prostacyclin, and tPA expression [58,67]. group and others that in ACS patients, local and circulating
CRP sustains an anti-inflammatory innate immune response T-clonotypes recruitment is selective and not random [76,77]
when circulating in a pentameric form, while as monomeric with the release of specific cytokines (Figure 1, Panel C)
form, it exerts potent proinflammatory and prothrombotic [47,83,86–88].
actions. Interestingly, this conversion may occur at the site A study from our group has clearly documented a specific,
of plaque rupture, mainly induced by activated platelets immune-driven response and inflammatory pathways within
[68,69]. the “culprit lesion” of patients presenting with ACS [75], show­
The role of IL-1β and related intra- and extracellular signal­ ing a specific oligoclonal T-cell expansion only in the vulner­
ing in all stages of atherosclerosis has been also reported able plaque and not in the peripheral blood, indicating
[70,71]. This interleukin induces expression of CAMs and a selective expansion driven by a given intraplaque antigen.
EXPERT REVIEW OF CARDIOVASCULAR THERAPY 355

To date, the putative antigens responsible for this intrale­ frontline host defense against microbial infection, and enhance
sion activation are still under investigation [29]. Modified lipo­ antigen presentation by APCs [104]. Moreover, platelets can
proteins, such as oxidized LDL (oxLDL), are thought to be the contribute to inflammation via direct secretion of proinflamma­
major determinant in T-cells activation via MCH-TCR interac­ tory mediators such as TGF-β1, -β2, -β4, IL-1α, IL-1β, IL-2-4, −6-8,
tion [89–91] or even the specific receptor for oxLDL, namely −10, −11, TNF-α, TNF-β, IFNγ, involved in the regulation of
LOX-1 [92]. In line with this hypothesis, we have recently angiogenesis, cellular proliferation and differentiation, chemo­
reported that oxLDL may induce T-cell activation in TCR inde­ taxis, vascular modeling, cellular interactions, and bone [105]. In
pendent manner inducing TF expression [93]. addition, upon activation through the expression of P-selectin,
In the last decade, researchers have focused more attention platelets rapidly bind to monocytes, forming monocyte–platelet
on a specific subset of Th-cells known as Th-17 that are able to aggregates (MPA) [107,108]. This interaction induces
release large amounts of IL-17, IL-21 granulocyte–macrophage a proinflammatory phenotype in circulating monocytes and
colony-stimulating factor, and IL-6 upon activation [94]. In vivo might be used in vivo as cellular biomarker of platelet activation
a small subset of CD161+ T-cell precursors may polarize into [109]. Of note, higher MPA levels have been reported both in
Th-17 cells upon IL-1β plus IL-23 stimulation. Their involve­ ACS patients and in those presenting with chronic coronary
ment in the pathogenesis of atherosclerosis is still matter of syndromes (CCS) [110,111], as well as in patients with endothe­
debate because of controversial reports. Some studies indicate lial dysfunction regardless of the presence of a coronary stenosis
that blocking IL-17 pathway may attenuate atherosclerosis [112]. Finally, different observations indicate a crosstalk between
progression [95–97], while other reports correlate IL-17 signal­ platelets and cancer: cancer cells may promote direct platelet
ing with plaque destabilization and acute event [98,99]. On aggregation or indirect platelet activation. These effect enhance
this regard, we have reported high levels of trans-coronary IL- the release of extracellular matrix proteins and TF from endothe­
17 and a higher number of Th-17 cells in a subgroup of ACS lial cells, thus favoring platelet adhesion and thrombus forma­
patients presenting with a more severe disease with higher tion [106]. On the other side, platelets store and release several
troponin levels at admission, as compared to stable angina angiogenic factors that balancing hemostasis and angiogenesis
patients. These data reinforce other studies on the possible within the tumor microenvironment finally contribute to cancer
contribution of Th-17 cells in determining a more severe dis­ angiogenesis, grow, and metastatic dissemination [106]. Taken
ease [98]. together, these observations give a newer point of view about
Moreover, we have shown that in vitro stimulation of platelet role in atherothrombosis, since they should not be
T-cells by a combination of IL-17 plus INF-γ may induce considered only as the final effector of the coagulation cascade
expression of TF with procoagulant activity [100]. TF positive but, on the contrary, as active players in the first steps of this
T-cells have been also found in vivo in coronary circulation disease.
as well as in thrombotic material aspirated from ACS Recently, a growing body of evidence is highlighting the
patients [100]. importance of miRNAs in atherosclerosis development and pro­
Finally, it has been postulated that the balance between gression specifically via the regulation of atherosclerosis-prone
plaque complication, vascular obstruction, myocardial ische­ genes [113]. miRNAs act by silencing their target genes, thus
mia, and occurrence of clinical events is in charge of Treg affecting cells function and protein synthesis [113]. Several
activity and its demodulation during acute myocardial infarc­ reports indicate their role in endothelial [114] and
tion may worse the outcome [101]. Further studies are war­ SMCs regulation [115–117] such as injury and consequent cell
ranted to better evaluate the role of Treg in CVD [102]. attachment, growth, and inflammatory responses [118]. On this
regard, another interesting chapter about the involvement of
miRNA in atherosclerosis and atherothrombosis is related to the
role of platelet miRNA [119]. Despite platelets do not have
4. Platelets in atherothrombosis: not only
nuclei, they contain several mRNAs [120] and rough endoplas­
aggregation
mic reticulum and polyribosomes, thus they have protein synth­
The physiological role of platelets in primary hemostasis is well esis (Figure 1, Panel B) [121]. We have reported that upon
known and studied [103]. Their main role is to prevent blood activation platelets are able to modify their proteome even in
loss when the vasculature is damaged [103]. The ‘classical’ point absence of substantial change in transcriptome [122]. These
of view about platelets in hemostasis has considered platelets modifications are part of an intense alternative splicing of
simply as the final effector of the coagulation cascade because mRNAs and miRNAs activity [123]. These small non-coding
they lack of nuclei [103]. However, newer evidence accumulated RNA interacts with a target mRNAs as a negative regulator,
in the last two decades have pointed out other novel functions thus down-regulating protein expression [124]. The potential
beyond aggregation because they seem able to modulate the role of Platelet-related miRNAs in the development of cardio­
immune response [104], to promote inflammation [105], and to vascular events is part of extensive research [125].
cause cancer metastasis [106]. Specifically, platelets have an
active role in innate immunity because they release several
5. ACS and functional alterations of coronary flow:
inflammatory and bioactive molecules stored within granules
beyond atherothrombosis
or synthesized upon activation [104]. These biochemical media­
tors, in turn, exert their effects by modulating the cells of the In about 10% of patients with ACS, myocardial ischemia is not
innate immune system. Moreover, platelets are directly involved due to atherothrombosis. In these cases, a functional altera­
as effector cells in innate immunity, have an active role in tion of coronary circulation involving both large epicardial
356 G. CIMMINO ET AL.

coronary arteries or the coronary microcirculation have been spasm. On the contrary, non-endothelium-dependent mechan­
indicated as responsible of the acute event [6]. The main role isms refer to structural remodeling of coronary microvasculature.
of the coronary circulation is to provide the cardiac muscle It consists in intimal thickening, perivascular fibrosis, capillary
with the oxygen it requires by regulating the coronary blood rarefaction with increased microvascular resistances finally
flow. In fact, myocardial oxygen extraction is almost maximal resulting in a CFR reduction [136]. Two different mechanisms
at rest, thereby an increase in oxygen demand would result in have been postulated to explain pathophysiology of MINOCA:
a corresponding increase in coronary blood flow. Coronary a. an acute trigger on top of a prolonged ischemia due to
microvascular disease (CMD) refers to the dysfunction of this a chronic microvascular dysfunction or, b. a sudden increase of
mechanism leading to myocardial ischemia and it is character­ resistance as consequence of an acute increase of adrenergic
ized by the presence of some abnormalities in the ‘function’ tone, as reported in Figure 2 [137]. Based on the pathogenesis
and ‘structure’ of the coronary microcirculation due to and in light of a tailored therapeutic approach, an invasive
endothelial and SMCs dysfunction. Most of the patients with assessment may plays a crucial role [137,138]. The first step is
CMD do not present ‘significant’ atherosclerotic obstructive ruling out hidden atherosclerotic causes such as acute plaque
coronary disease at coronary angiography despite presenting rupture/erosion or the presence of functionally significant and
with typical chest pain. angiographically underestimated intermediate coronary artery
At the end of 1999 the WISE study was published reporting stenosis, as shown in Table 1. IVUS and OCT are useful to assess
an abnormal coronary flow reserve (CFR) in half of women the quality of atherosclerotic plaques among the epicardial cor­
with suspected ischemic heart disease but with non- onary circulation, identifying signs of plaque disruption or cor­
obstructive coronary arteries [126]. Similarly, other studies onary dissection, while FFR allows functional assessment of
showed that approximately 50% of patients presenting with angiographically intermediate coronary stenosis ruling out the
myocardial infarction with non-obstructive coronary arteries presence of hemodynamically significant obstructive CAD. Once
(MINOCA) experience CMD mainly associated with microvas­ ruling out atherosclerotic causes of MINOCA, both thermodilu­
cular spasm [127–131]. tion-based assessment of coronary microvascular function and
A previous meta-analysis of 28 studies showed that provocative test with acetylcholine can be performed. The for­
MINOCA is relatively common accounting for 6% of all myo­ mer evaluates microvascular vasodilatation by measuring both
cardial infarctions (MI) [128] .Females are more common than CFR and index of microvascular resistance (IMR), the latter is
men and patients are slightly younger as compared with MI useful to rule out endothelial dysfunction presenting with para­
associated with CAD [128,132]. MINOCA patients were initially doxical vasoconstriction, induced by intracoronary acetylcholine
considered to have a benign prognosis, however, recent stu­ injection. In fact, in case of intact endothelium, endothelium-
dies and metanalysis have shown an increased risk of both dependent vasodilation normally opposes to α-adrenergic vaso­
new cardiovascular events and hospitalization for heart failure constriction, while at the early stage of the atherosclerotic
and stroke with a mortality rate of 2% per annum [132–134]. process a constrictor response emerges [139]. Of note, in subjects
In the pathogenesis of CMD are involved both endothelium- with normal coronary angiograms, there is no α-adrenergic cor­
dependent and non-endothelium-dependent mechanisms. onary constrictor tone at rest and the constrictor response during
Endothelium-dependent mechanisms refer to the endothelial sympathetic activation is mediated by α2-adrenoceptors and
dysfunction, which is responsible of a dysregulation of the micro­ mainly affects the microcirculation. In patients with endothelial
vascular reactivity, affecting the physiological flow-mediated dysfunction, sympathetic activation leads to an enhanced α-
vasodilation that is needed when an increased myocardial oxy­ adrenergic vasoconstriction possibly leading to myocardial
gen demand occur [135]. This final event may even result in ischemia in atherosclerotic patients, in whom the α2-adrenergic
a paradoxical vasoconstriction, thus producing microvascular coronary constriction tone is enhanced [139].

Figure 2. Pathophysiology of Myocardial infarction in non-obstructive CAD. The most common causes of MINOCA are represented by coronary plaque disease,
coronary dissection, coronary artery spasm, coronary microvascular dysfunction, and coronary thromboembolism.
EXPERT REVIEW OF CARDIOVASCULAR THERAPY 357

Another alternative cause of CMD may be coronary micro­ 6. Conclusions


embolization [140]. Microemboli usually consist of material
The current guidelines simplify the ACS based on ECG mod­
coming from the atherosclerotic plaque, including cholesterol,
ifications: ST-segment myocardial infarction versus non-ST-
platelet and platelets-leukocyte aggregates, and fibrin [141–
segment myocardial infarction. However, in the last few
145]. Microembolization induces patchy microinfarcts asso­
years, this simplified clinical view has been associated in par­
ciated with a typical time-course of the regional contractile
allel to a complex pathophysiological scenario since not all
function which is characterized by an immediate decrease
cases of ACS fit the coronary stenosis substrate. As we have
followed by a partial recovery and a subsequent deterioration
discussed in the present manuscript, we need to move
[146,147]. Several studies have indicated that inflammation
beyond atherosclerotic plaque-related stenosis considering
might play a pivotal role in this phenomenon. Specifically,
arterial spasm, microvascular dysfunction, rupture without
experimental studies have shown a significant unbalance of
inflammation, and immunity involvement as instigators of
expression of cytokines with pro and anti-inflammatory role in
plaque erosion.
coronary microembolization (9). Among them, TNF-alpha
Control of traditional risk factors remains essential in pre­
appears as the pivotal molecule because it is up expressed,
venting ACS occurrence, but the role of inflammation, immu­
in both macrophages and cardiomyocytes, and it is linked to
nity, vasospasm, and microvascular dysfunction should also be
a negative inotropic effect [148–150]. Coronary microemboli­
considered especially for the less well-recognized pathways to
zation may occur either spontaneously or because of percuta­
acute myocardial ischemia discussed in the present review.
neous coronary interventions (PCI). In the context of a stable
Newer biomarkers (such as miRNAs) or application of novel
clinical setting, coronary microembolization can be easily iden­
imaging techniques may represent the challenge in the triage
tified after PCI where a transient increase of high-sensitive
of ACS patients. Based on the ACS mortality and morbidity
troponins identifies the occurrence of a peri-procedural myo­
rate, we cannot to be satisfied with our current approach to
cardial injury [151]. However, the elevation in troponin levels
ACS patients. We are maybe in need of a more personalized
after PCI can be associated with either a side-branch occlu­
precision deployment of treatment strategies with a look to
sion/damage or distal coronary microembolization. On the
the diverse underlying causes of acute myocardial ischemia.
contrary, during ACS, it can be difficult to distinguish whether
the increase in cardiac biomarker levels is mainly due to
spontaneous coronary microembolization rather than other
7. Expert opinion
causes. In this setting, microcirculation might also be exposed
to myocardial ischemia/reperfusion injury. The no-reflow phe­ In the last few years, despite the great advancements in our
nomenon is the practical example of this damage as it may understanding of ACS pathophysiology, ischemic cardiovascular
occur after primary PCI, particularly after thrombolysis (rescue diseases and their complications remain the major cause of mor­
PCI), when soon after revascularization of the occluded cor­ bidity and mortality in western countries. Atherothrombosis
onary artery there is slow- or even no-flow downstream the accounts almost the majority of ACS, while a small contribution
vessel [152,153]. Obstructed microcirculation can be damaged comes from the non-obstructive coronary artery forms. Although
by reversible edema eventually associated with capillary it has been clearly indicated that several mediators of inflamma­
destruction and intramyocardial hemorrhage [152,154–156]. tion and many cell types belonging to the immunological system
Microcirculation protection, including different strategies
are actively involved in the development of stable atherosclerotic
ranges from pre- or postconditioning, remote ischemia and
plaques and their instability, the relative role of single cell type and
hypothermia, represents the new frontiers of the modern
their reciprocal role in the network of immunity are not completely
cardiology.
understood yet. Moreover, the identification of possible antigens
According to the Coronary Vasomotion Disorders International
responsible of activation of cells of immunity is a still unexplored
Study Group (COVADIS) indications, during acetylcholine test, the
presence of both typical symptoms (usually experienced by the area with many questions without answers. This is a crucial area of
patients) and ischemic ECG changes, in the absence of significative interest because identification of these antigens might be consid­
epicardial spasm, allow the diagnosis of CMD associated with ered as the first step to study newer potential therapeutical tar­
microvascular vasospasm [131]. However, CMD cannot be consid­ gets. The hypothesis of a “vaccine” against ACS might be
ered only an isolated disease, since it might be discovered also in a fascinating drive to stimulate the research in this field. Thus,
patients presenting with CCS. In addition, CMD can also be a look into the future of therapy of ACS might consider not only
a consequence of myocardial injury either associated with coron­ drugs against lipids and thrombosis but also drugs to obtain fine
ary revascularization or others ischemic or nonischemic causes modulation of immune-inflammation in order to avoid not only
[157–161]. It has been found to be strongly associated with plaques grow but also their conversion from ‘stable’ to ‘unstable.’
heart failure with preserved ejection fraction, diabetes, hyperten­ This complex pathophysiological scenario is not applicable
sion, and it also confers an increased risk of adverse cardiovascular in the ACS in which a non-obstructive CAD (MINOCA) is docu­
events [162]. mented during the acute event. Since this is another largely
In conclusions, ACS are complex clinical syndromes with unexplored area, the currently available tools in the cath-lab
the involvement of different pathophysiological mechanisms, with the assessment of both CFR and IMR together with
including vasomotion abnormalities and coronary microvascu­ provocative tests will provide an improvement of our knowl­
lar dysfunction. The latter represent the main mechanisms edge of the pathophysiology of this condition in order to
associated with the occurrence of MINOCA. provide the patients with the best tailored therapy.
358 G. CIMMINO ET AL.

Prevention of new onset or recurrent coronary events is the 6. Crea F, Libby P. Acute coronary syndromes: the way forward from
challenge of the next future. The great improvement on the mechanisms to precision treatment. Circulation. 2017 Sep 19;136
(12):1155–1166.
application of novel imaging and molecular techniques might
•• this is an extensive review in the field
represent the opportunity to early identify the patient at risk to 7. Abdu FA, Mohammed A-Q, Liu L, et al. Myocardial Infarction with
develop an ACS and better characterize the specific pattern Nonobstructive Coronary Arteries (MINOCA): a review of the cur­
involved in the acute as well as the chronic phase. Based on the rent position. Cardiology. 2020;145(9):543–552.
current knowledge, immune-inflammatory modulation using spe­ 8. Lowe GD. Local inflammation, endothelial dysfunction and fibrino­
lysis in coronary heart disease. Clin Sci. 2006 Mar;110(3):327–328.
cific drugs might be of interest. Identification of the possible
9. Grassi D, Desideri G, Ferri C. Cardiovascular risk and endothelial
antigens involved in the immune system activation within athero­ dysfunction: the preferential route for atherosclerosis. Curr Pharm
sclerotic lesion might shed more light to pursue new and specific Biotechnol. 2011 Sep;12(9):1343–1353.
therapeutical targets. On the other hand, the management of 10. Grover-Paez F, Zavalza-Gomez AB. Endothelial dysfunction and
non-obstructive CAD remains to be better defined. Finally, the cardiovascular risk factors. Diabetes Res Clin Pract. 2009 Apr;84
(1):1–10.
discovery of novel roles for the platelets beyond the simplified
11. Jowett JB. Interplay of genetic and environmental factors: innate
aggregation process represent another important finding to immunity genetic polymorphisms in MBL2 affect endothelial dys­
further investigate. function and risk of atherosclerosis. Atherosclerosis. 2010 Jan;208
(1):32–33.
12. Santos-Gallego CG, Badimon JJ, Rosenson RS. Beginning to under­
Declaration of Interest stand high-density lipoproteins. Endocrinol Metab Clin North Am.
2014 Dec;43(4):913–947.
The authors have no relevant affiliations or financial involvement with any
13. Hutter R, Speidl WS, Valdiviezo C, et al. Macrophages transmit
organization or entity with a financial interest in or financial conflict with
potent proangiogenic effects of oxLDL in vitro and in vivo
the subject matter or materials discussed in the manuscript. This includes
involving HIF-1alpha activation: a novel aspect of angiogenesis
employment, consultancies, honoraria, stock ownership or options, expert
in atherosclerosis. J Cardiovasc Transl Res. 2013 Aug;6
testimony, grants or patents received or pending, or royalties.
(4):558–569.
14. Boren J, Chapman MJ, Krauss RM, et al. Low-density lipoproteins
cause atherosclerotic cardiovascular disease: pathophysiological,
Reviewer disclosures genetic, and therapeutic insights: a consensus statement from
Peer reviewers on this manuscript have no relevant financial or other the European Atherosclerosis Society Consensus Panel. Eur Heart
relationships to disclose. J. 2020 Jun 21;41(24):2313–2330.
15. Libby P. The changing landscape of atherosclerosis. Nature. 2021
Apr;592(7855):524–533.
Author contributions 16. Khalil MF, Wagner WD, Goldberg IJ. Molecular interactions leading
to lipoprotein retention and the initiation of atherosclerosis.
All authors equally contributed to this paper with conception and design Arterioscler Thromb Vasc Biol. 2004 Dec;24(12):2211–2218.
of the study, literature review and analysis, drafting and critical revision 17. Wolf D, Ley K. Immunity and Inflammation in Atherosclerosis. Circ
and editing, and final approval of the final version. Res. 2019 Jan 18;124(2):315–327.
• this is an interesting paper exploring the role of immunty in
atherosclerosis
Funding 18. Riksen NP, Stienstra R. Metabolism of innate immune cells: impact
on atherosclerosis. Curr Opin Lipidol. 2018 Oct;29(5):359–367.
This paper funded by V:ALERE 2019 grant from University of Campania
19. Ruuth M, Nguyen SD, Vihervaara T, et al. Susceptibility of
Luigi Vanvitelli.
low-density lipoprotein particles to aggregate depends on particle
lipidome, is modifiable, and associates with future cardiovascular
deaths. Eur Heart J. 2018 Jul 14;39(27):2562–2573.
ORCID 20. Llorente-Cortés V, Martínez-González J, Badimon L. LDL receptor–
Plinio Cirillo http://orcid.org/0000-0001-7818-4952 related protein mediates uptake of aggregated LDL in human
vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2000
Jun;20(6):1572–1579.
References 21. Bennett MR, Sinha S, Owens GK. Vascular smooth muscle cells in
atherosclerosis. Circ Res. 2016 Feb 19;118(4):692–702.
Papers of special note have been highlighted as either of interest (•) or of 22. Arbustini E, Grasso M, Diegoli M, et al. Coronary atherosclerotic
considerable interest (••) to readers. plaques with and without thrombus in ischemic heart syndromes:
1. Badimon JJ, Ibanez B, Cimmino G. Genesis and dynamics of ather­ a morphologic, immunohistochemical, and biochemical study. Am
osclerotic lesions: implications for early detection. Cerebrovascular J Cardiol. 1991 Sep 3;68(7):36B–50B.
Dis. 2009;27(1):38–47. 23. Kim Y, Kim B-K, Johnson TW, et al. Silent plaque rupture in the left
2. Cimmino G, Conte S, Morello A, et al. The complex puzzle under­ main stem assessed by optical coherence tomography. Cardiol J.
lying the pathophysiology of acute coronary syndromes: from 2020;27(3):316–317.
molecular basis to clinical manifestations. Expert Rev Cardiovasc 24. Lutgens E, van Suylen R-J, Faber BC, et al. Atherosclerotic plaque
Ther. 2012 Dec;10(12):1533–1543. rupture: local or systemic process? Arterioscler Thromb Vasc Biol.
• this is an extensive review in the field 2003 Dec;23(12):2123–2130.
3. Weber C, Noels H. Atherosclerosis: current pathogenesis and ther­ 25. Ford TJ, Corcoran D, Berry C. Stable coronary syndromes:
apeutic options. Nat Med. 2011;17(11):1410–1422. pathophysiology, diagnostic advances and therapeutic need.
4. Ross R, Epstein FH. Atherosclerosis — an inflammatory disease. Heart (British Cardiac Society). 2018 Feb;104(4):284–292.
N Engl J Med. 1999 Jan 14;340(2):115–126. 26. Cimmino G, Loffredo FS, Morello A, et al. Immune-Inflammatory
5. Meerarani P, Moreno PR, Cimmino G, et al. Atherothrombosis: role Activation in Acute Coronary Syndromes: a Look into the Heart
of tissue factor; link between diabetes, obesity and inflammation. of Unstable Coronary Plaque. Curr Cardiol Rev. 2017;13(2):110
Indian J Exp Biol. 2007 Jan;45(1):103–110. –117.
EXPERT REVIEW OF CARDIOVASCULAR THERAPY 359

27. Cimmino G, Cirillo P. Tissue factor: newer concepts in thrombosis 49. Libby P, Ridker PM. Inflammation and atherosclerosis: role of
and its role beyond thrombosis and hemostasis. Cardiovasc Diagn C-reactive protein in risk assessment. Am J Med. 2004 Mar 22;
Ther. 2018 Oct;8(5):581–593. Suppl 116(6):9S–16S.
28. Hansson GK, Hermansson A. The immune system in atherosclerosis. 50. Michowitz Y, Arbel Y, Wexler D, et al. Predictive value of high
Nat Immunol. 2011 Mar;12(3):204–212. sensitivity CRP in patients with diastolic heart failure. Int J Cardiol.
29. Ketelhuth DF, Hansson GK. Cellular immunity, low-density lipopro­ 2008 Apr 25;125(3):347–351.
tein and atherosclerosis: break of tolerance in the artery wall. 51. Niccoli G, Biasucci LM, Biscione C, et al. Independent prognostic value
Thromb Haemost. 2011 Nov;106(11):779–786. of C-reactive protein and coronary artery disease extent in patients
30. Libby P, Ridker PM, Hansson GK. Progress and challenges in trans­ affected by unstable angina. Atherosclerosis. 2008 Feb;196(2):779–785.
lating the biology of atherosclerosis. Nature. 2011 May 19;473 52. Zamani P, Schwartz GG, Olsson AG, et al. Inflammatory biomarkers,
(7347):317–325. death, and recurrent nonfatal coronary events after an acute cor­
31. Michel J-B, Martin-Ventura JL, Nicoletti A, et al. Pathology of onary syndrome in the MIRACL study. J Am Heart Assoc. 2013 Jan
human plaque vulnerability: mechanisms and consequences of 28;2(1):e003103.
intraplaque haemorrhages. Atherosclerosis. 2014 Jun;234 53. Ridker PM, Thuren T, Zalewski A, et al. Interleukin-1beta inhibition
(2):311–319. and the prevention of recurrent cardiovascular events: rationale
32. Hansson GK, Libby P, Tabas I. Inflammation and plaque and design of the Canakinumab Anti-inflammatory Thrombosis
vulnerability. J Intern Med. 2015 Aug 11;278(5):483–493. Outcomes Study (CANTOS). Am Heart J. 2011 Oct;162(4):597–605.
33. Libby P. Inflammation in atherosclerosis. Nature. 2002 Dec 19- 54. Date H, Imamura T, Sumi T, et al. Effects of interleukin-6 produced in
26;420(6917):868–874. coronary circulation on production of C-reactive protein and coronary
34. Stefanadis C, Antoniou C-K, Tsiachris D, et al. Coronary athero­ microvascular resistance. Am J Cardiol. 2005 Apr 1;95(7):849–852.
sclerotic vulnerable plaque: current perspectives. J Am Heart 55. Tsuda K. C-reactive protein and nitric oxide production in ischemic
Assoc. 2017 Mar 17;6(3). 10.1161/JAHA.117.005543. stroke. author reply e472. Stroke. 2009 Jun;40(6):e471.
35. Hafiane A. Vulnerable plaque, characteristics, detection, and poten­ 56. Forte L, Cimmino G, Loffredo F, et al. C-reactive protein is released in the
tial therapies. J Cardiovasc Dev Dis. 2019 Jul 27;6(3):26. coronary circulation and causes endothelial dysfunction in patients
36. Ezekowitz JA, Bakal JA, Westerhout CM, et al. The relationship with acute coronary syndromes. Int J Cardiol. 2011 Oct 6;152(1):7–12.
between meteorological conditions and index acute coronary 57. Montecucco F, Steffens S, Burger F, et al. C-reactive protein (CRP)
events in a global clinical trial. Int J Cardiol. 2013 Oct 3;168 induces chemokine secretion via CD11b/ICAM-1 interaction in human
(3):2315–2321. adherent monocytes. J Leukoc Biol. 2008 Oct;84(4):1109–1119.
37. Gasparrini A, Guo Y, Hashizume M, et al. Mortality risk attributable 58. Pasceri V, Willerson JT, Yeh ET. Direct proinflammatory effect of
to high and low ambient temperature: a multicountry observa­ C-reactive protein on human endothelial cells. Circulation. 2000
tional study. Lancet. 2015 Jul 25;386(9991):369–375. Oct 31;102(18):2165–2168.
38. Wolf K, Schneider A, Breitner S, et al. Air temperature and the 59. Portelinha A, Belo L, Tejera E, et al. Adhesion molecules (VCAM-1
occurrence of myocardial infarction in Augsburg, Germany. and ICAM-1) and C-reactive protein in women with history of
Circulation. 2009 Sep 1;120(9):735–742. preeclampsia. Acta Obstet Gynecol Scand. 2008;87(9):969–971.
39. Saeki K, Obayashi K, Iwamoto J, et al. The relationship between 60. Postadzhiyan AS, Tzontcheva AV, Kehayov I, et al. Circulating soluble
indoor, outdoor and ambient temperatures and morning BP surges adhesion molecules ICAM-1 and VCAM-1 and their association with
from inter-seasonally repeated measurements. J Hum Hypertens. clinical outcome, troponin T and C-reactive protein in patients with
2014 Aug;28(8):482–488. acute coronary syndromes. Clin Biochem. 2008 Feb;41(3):126–133.
40. Eldwood PC, Beswick A, O’Brien JR, et al. Temperature and risk 61. Cirillo P, Golino P, Calabro P, et al. C-reactive protein induces tissue
factors for ischaemic heart disease in the Caerphilly prospective factor expression and promotes smooth muscle and endothelial
study. Br Heart J. 1993 Dec;70(6):520–523. cell proliferation. Cardiovasc Res. 2005 Oct 1;68(1):47–55.
41. Neild PJ, Syndercombe-Court D, Keatinge WR, et al. Cold-induced 62. Guo S, Meng S, Chen B, et al. C-reactive protein can influence the
increases in erythrocyte count, plasma cholesterol and plasma proliferation, apoptosis, and monocyte chemotactic protein-1 pro­
fibrinogen of elderly people without a comparable rise in protein duction of human umbilical vein endothelial cells. DNA Cell Biol.
C or factor X. Clin Sci. 1994 Jan;86(1):43–48. 2011 Mar;30(3):157–162.
42. Raphael CE, Heit JA, Reeder GS, et al. Coronary embolus: an under­ 63. Cimmino G, Ragni M, Cirillo P, et al. C-reactive protein induces
appreciated cause of acute coronary syndromes. JACC: Cardiovascular expression of matrix metalloproteinase-9: a possible link between
Interventions. 2018 Jan 22;11(2):172–180. inflammation and plaque rupture. Int J Cardiol. 2012 Nov 14.
43. Kolodgie FD, Virmani R, Finn AV, et al. Embolic myocardial infarc­ 10.1016/j.ijcard.2012.10.040.
tion as a consequence of atrial fibrillation: a prevailing disease of 64. Gresele P, Falcinelli E, Loffredo F, et al. Platelets release matrix
the future. Circulation. 2015 Jul 28;132(4):223–226. metalloproteinase-2 in the coronary circulation of patients with
44. Liu G, Yang P, He Y. Left ventricular mural thrombus and dual acute coronary syndromes: possible role in sustained platelet
coronary embolization associated with hyperthyroid cardiomyopa­ activation. Eur Heart J. 2011 Feb;32(3):316–325.
thy and atrial fibrillation: a case report. BMC Cardiovasc Disord. 65. Galve-de Rochemonteix B, Wiktorowicz K, Kushner I, et al.
2017 May 19;17(1):128. C-reactive protein increases production of IL-1α, IL-1β, and TNF-α,
45. Lacunza-Ruiz FJ, Munoz-Esparza C, Garcia-de-Lara J. Coronary and expression of mRNA by human alveolar macrophages.
embolism and thrombosis of prosthetic mitral valve. J Leukoc Biol. 1993 Apr;53(4):439–445.
JACC: Cardiovascular Interventions. 2014 Oct;7(10):e127–8. 66. Xie L, Chang L, Guan Y, et al. C-reactive protein augments
46. Garachemani A, Eshtehardi P, Meier B. Paradoxical emboli through interleukin-8 secretion in human peripheral blood monocytes.
the patent foramen ovale as the suspected cause of myocardial J Cardiovasc Pharmacol. 2005 Nov;46(5):690–696.
and renal infarction in a 48-year-old woman. Catheter Cardiovasc 67. Hattori Y, Matsumura M, Kasai K. Vascular smooth muscle cell activation
Interv. 2007 Dec 1;70(7):1010–1012. by C-reactive protein. Cardiovasc Res. 2003 Apr 1;58(1):186–195.
47. Correia LCL, Andrade BB, Borges VM, et al. Prognostic value of 68. Eisenhardt SU, Habersberger J, Murphy A, et al. Dissociation of
cytokines and chemokines in addition to the GRACE Score in pentameric to monomeric C-reactive protein on activated platelets
non-ST-elevation acute coronary syndromes. Clin Chim Acta. 2010 localizes inflammation to atherosclerotic plaques. Circ Res. 2009 Jul
Apr 2;411(7–8):540–545. 17;105(2):128–137.
48. Li -J-J, Jiang H, Huang C-X, et al. Elevated level of plasma 69. Thiele JR, Habersberger J, Braig D, et al. Dissociation of pentameric to
C-reactive protein in patients with unstable angina: its relations monomeric C-reactive protein localizes and aggravates inflammation:
with coronary stenosis and lipid profile. Angiology. 2002 May- in vivo proof of a powerful proinflammatory mechanism and a new
Jun;53(3):265–272. anti-inflammatory strategy. Circulation. 2014 Jul 1;130(1):35–50.
360 G. CIMMINO ET AL.

70. Mai W, Liao Y. Targeting IL-1β in the Treatment of Atherosclerosis. activation of lectin-like oxidized low-density lipoprotein
Front Immunol. 2020;11:589654. receptor-1. Cardiovasc Res. 2020 May 1;116(6):1125–1135.
71. Vromman A, Ruvkun V, Shvartz E, et al. Stage-dependent differential 94. Taleb S, Tedgui A, Mallat Z. IL-17 and Th17 cells in atherosclerosis:
effects of interleukin-1 isoforms on experimental atherosclerosis. Eur subtle and contextual roles. Arterioscler Thromb Vasc Biol. 2015
Heart J. 2019 Aug 7;40(30):2482–2491. Feb;35(2):258–264.
72. Dinarello CA. Immunological and inflammatory functions of the 95. Erbel C, Chen L, Bea F, et al. Inhibition of IL-17A attenuates athero­
interleukin-1 family. Annu Rev Immunol. 2009;27(1):519–550. sclerotic lesion development in apoE-deficient mice. The Journal of
73. Su J-H, Luo M-Y, Liang N, et al. Interleukin-6: a novel target for Immunology. 2009 Dec 15;183(12):8167–8175.
cardio-cerebrovascular diseases. Front Pharmacol. 2021;12:745061. 96. Taleb S, Romain M, Ramkhelawon B, et al. Loss of SOCS3 expression
74. Epelman S, Mann DL. Communication in the heart: the role of the in T cells reveals a regulatory role for interleukin-17 in
innate immune system in coordinating cellular responses to atherosclerosis. J Exp Med. 2009 Sep 28;206(10):2067–2077.
ischemic injury. J Cardiovasc Transl Res. 2012 Dec;5(6):827–836. 97. Gistera A, Robertson A-KL, Andersson J, et al. Transforming growth
75. De Palma R, Del Galdo F, Abbate G, et al. Patients with acute factor–β signaling in T cells promotes stabilization of atherosclero­
coronary syndrome show oligoclonal T-cell recruitment within tic plaques through an interleukin-17–dependent pathway. Sci
unstable plaque: evidence for a local, intracoronary immunologic Transl Med. 2013 Jul 31;5(196):196ra100.
mechanism. Circulation. 2006 Feb 7;113(5):640–646. 98. Cirillo P, Golino P, Piscione F, et al. Transcoronary Th-17 lympho­
•• This is an important article in the role of immunity in vulner­ cytes and acute coronary syndromes: new evidence from the crime
able plaque scene? Int J Cardiol. 2011 Dec 1;153(2):215–216.
76. Liuzzo G, Goronzy JJ, Yang H, et al. Monoclonal T-cell proliferation 99. Erbel C, Dengler TJ, Wangler S, et al. Expression of IL-17A in human
and plaque instability in acute coronary syndromes. Circulation. atherosclerotic lesions is associated with increased inflammation
2000 Jun 27;101(25):2883–2888. and plaque vulnerability. Basic Res Cardiol. 2011 Jan;106
77. Liuzzo G, Kopecky SL, Frye RL, et al. Perturbation of the T-cell (1):125–134.
repertoire in patients with unstable angina. Circulation. 1999 Nov 100. De Palma R, Cirillo P, Ciccarelli G, et al. Expression of functional
23;100(21):2135–2139. tissue factor in activated T-lymphocytes in vitro and in vivo:
78. Wang C, Jin R, Zhu X, et al. Function of CD147 in atherosclerosis a possible contribution of immunity to thrombosis? Int J Cardiol.
and atherothrombosis. J Cardiovasc Transl Res. 2015 Feb;8 2016 Sep 1;218:188–195.
(1):59–66. 101. Caligiuri G, Nicoletti A. Lymphocyte responses in acute coronary
79. Moore KJ, Tabas I. Macrophages in the pathogenesis of syndromes: lack of regulation spawns deviant behaviour. Eur Heart
atherosclerosis. Cell. 2011 Apr 29;145(3):341–355. J. 2006 Nov;27(21):2485–2486.
80. Hedrick CC. Lymphocytes in atherosclerosis. Arterioscler Thromb 102. Albany CJ, Trevelin SC, Giganti G, et al. Getting to the heart of the
Vasc Biol. 2015 Feb;35(2):253–257. matter: the role of regulatory T-cells (tregs) in cardiovascular dis­
81. Tse K, Tse H, Sidney J, et al. T cells in atherosclerosis. Int Immunol. ease (CVD) and atherosclerosis. Front Immunol. 2019;10:2795.
2013 Nov;25(11):615–622. 103. Cimmino G, Golino P. Platelet biology and receptor pathways.
82. Doherty TM. T-cell regulation of macrophage function. Curr Opin J Cardiovasc Transl Res. 2013 Jun;6(3):299–309.
Immunol. 1995 Jun;7(3):400–404. 104. Ali RA, Wuescher LM, Worth RG. Platelets: essential components of
83. Profumo E, Buttari B, Tosti ME, et al. Plaque-infiltrating the immune system. Curr Trends Immunol. 2015;16:65–78.
T lymphocytes in patients with carotid atherosclerosis: an insight 105. Nurden AT. The biology of the platelet with special reference to
into the cellular mechanisms associated to plaque destabilization. inflammation, wound healing and immunity. Front Biosci. 2018 Jan
J Cardiovasc Surg (Torino). 2013 Jun;54(3):349–357. 1;23(2):726–751.
84. Huber SA, Sakkinen P, David C, et al. T helper–cell phenotype 106. Braun A, Anders HJ, Gudermann T, et al. Platelet-cancer interplay:
regulates atherosclerosis in mice under conditions of mild hyperch­ molecular mechanisms and new therapeutic avenues. Front Oncol.
olesterolemia. Circulation. 2001 May 29;103(21):2610–2616. 2021;11:665534.
85. Caligiuri G, Paulsson G, Nicoletti A, et al. Evidence for 107. Danenberg HD, Kantak N, Grad E, et al. C-reactive protein promotes
antigen-driven T-cell response in unstable angina. Circulation. monocyte-platelet aggregation: an additional link to the
2000 Sep 5;102(10):1114–1119. inflammatory-thrombotic intricacy. Eur J Haematol. 2007 Mar;78
86. Chistiakov DA, Orekhov AN, Bobryshev YV. Immune-inflammatory (3):246–252.
responses in atherosclerosis: role of an adaptive immunity mainly 108. Kälsch T, Elmas E, Nguyen XD, et al. Endotoxin-induced effects on
driven by T and B cells. Immunobiology. 2016 Sep;221(9): platelets and monocytes in an in vivo model of inflammation. Basic
1014–1033. Res Cardiol. 2007 Sep;102(5):460–466.
87. Cirillo P, Cimmino G, D’Aiuto E, et al. Local cytokine production in 109. Passacquale G, Vamadevan P, Pereira L, et al. Monocyte-platelet
patients with Acute Coronary Syndromes: a look into the eye of the interaction induces a pro-inflammatory phenotype in circulating
perfect (cytokine) storm. Int J Cardiol. 2014 Sep;176(1):227–229. monocytes. PLoS One. 2011;6(10):e25595.
88. Cheng X, Liao Y-H, Ge H, et al. TH1/TH2 functional imbalance after 110. Furman MI, Benoit SE, Barnard MR, et al. Increased platelet reactiv­
acute myocardial infarction: coronary arterial inflammation or myo­ ity and circulating monocyte-platelet aggregates in patients with
cardial inflammation. J Clin Immunol. 2005 May;25(3):246–253. stable coronary artery disease. J Am Coll Cardiol. 1998 Feb;31
89. Quinn MT, Parthasarathy S, Fong LG, et al. Oxidatively modified low (2):352–358.
density lipoproteins: a potential role in recruitment and retention 111. Hamilos M, Sarma J, Ostojic M, et al. Interference of drug-eluting
of monocyte/macrophages during atherogenesis. Proc Natl Acad stents with endothelium-dependent coronary vasomotion: evi­
Sci U S A. 1987 May;84(9):2995–2998. dence for device-specific responses. Circ Cardiovasc Interv. 2008
90. Stemme S, Faber B, Holm J, et al. T lymphocytes from human Dec;1(3):193–200.
atherosclerotic plaques recognize oxidized low density 112. Di Serafino L, Sarma J, Dierickx K, et al. Monocyte-platelets aggre­
lipoprotein. Proc Natl Acad Sci U S A. 1995 Apr 25;92(9):3893–3897. gates as cellular biomarker of endothelium- dependent coronary
91. Li D, Mehta JL. Oxidized LDL, a critical factor in atherogenesis. vasomotor dysfunction in patients with coronary artery disease.
Cardiovasc Res. 2005 Dec 1;68(3):353–354. J Cardiovasc Transl Res. 2014;7(1):1–8.
92. Di Pietro N, Formoso G, Pandolfi A. Physiology and pathophysiol­ 113. Lu Y, Thavarajah T, Gu W, et al. Impact of miRNA in Atherosclerosis.
ogy of oxLDL uptake by vascular wall cells in atherosclerosis. Vascul Arterioscler Thromb Vasc Biol. 2018 Sep;38(9):e159–e170.
Pharmacol. 2016 Sep;84:1–7. 114. Fernandez-Hernando C, Suarez Y. MicroRNAs in endothelial cell
93. Cimmino G, Cirillo P, Conte S, et al. Oxidized low-density lipopro­ homeostasis and vascular disease. Curr Opin Hematol. 2018
teins induce tissue factor expression in T-lymphocytes via May;25(3):227–236.
EXPERT REVIEW OF CARDIOVASCULAR THERAPY 361

115. Reddy MA, Das S, Zhuo C, et al. Regulation of vascular smooth 136. Pries AR, Badimon L, Bugiardini R, et al. Coronary vascular regula­
muscle cell dysfunction under diabetic conditions by miR-504. tion, remodelling, and collateralization: mechanisms and clinical
Arterioscler Thromb Vasc Biol. 2016 May;36(5):864–873. implications on behalf of the working group on coronary patho­
116. Albinsson S, Sward K. Targeting smooth muscle microRNAs for ther­ physiology and microcirculation. Eur Heart J. 2015 Dec 01;36
apeutic benefit in vascular disease. Pharmacol Res. 2013 (45):3134–3146.
Sep;75:28–36. 137. Mangiacapra F, Viscusi MM, Paolucci L, et al. The pivotal role of
117. Robinson HC, Baker AH. How do microRNAs affect vascular smooth invasive functional assessment in patients with myocardial infarc­
muscle cell biology? Curr Opin Lipidol. 2012 Oct;23(5):405–411. tion with non-obstructive coronary arteries (MINOCA). Front
118. Hergenreider E, Heydt S, Treguer K, et al. Atheroprotective com­ Cardiovasc Med. 2021;8:781485.
munication between endothelial cells and smooth muscle cells 138. Lindahl B, Baron T, Albertucci M, et al. Myocardial infarction with
through miRNAs. Nat Cell Biol. 2012 Feb 12;14(3):249–256. non-obstructive coronary artery disease. EuroIntervention. 2021
119. Yun SH, Sim EH, Goh RY, et al. Platelet activation: the mechanisms Dec 03;17(11):e875–e887.
and potential biomarkers. Biomed Res Int. 2016;2016:9060143.
139. Heusch G, Baumgart D, Camici P, et al. alpha-adrenergic coronary
120. Gnatenko DV, Dunn JJ, McCorkle SR, et al. Transcript profiling of
vasoconstriction and myocardial ischemia in humans. Circulation.
human platelets using microarray and serial analysis of gene
2000 Feb 15;101(6):689–694.
expression. Blood. 2003 Mar 15;101(6):2285–2293.
140. Kleinbongard P, Heusch G. A fresh look at coronary
121. Kieffer N, Guichard J, Farcet JP, et al. Biosynthesis of major platelet
microembolization. l. 2022 Apr;19(4):265–280.
proteins in human blood platelets. Eur J Biochem. 1987 Apr 1;164
(1):189–195. 141. Falk E. Plaque rupture with severe pre-existing stenosis precipitat­
122. Cimmino G, Tarallo R, Nassa G, et al. Activating stimuli induce ing coronary thrombosis. Characteristics of coronary atherosclerotic
platelet microRNA modulation and proteome reorganisation. plaques underlying fatal occlusive thrombi. Br Heart J. 1983 Aug;50
Thromb Haemost. 2015 Jul;114(1):96–108. (2):127–134.
123. Nassa G, Giurato G, Cimmino G, et al. Splicing of platelet resident 142. Falk E. Unstable angina with fatal outcome: dynamic coronary
pre-mRNAs upon activation by physiological stimuli results in function­ thrombosis leading to infarction and/or sudden death. Autopsy
ally relevant proteome modifications. Sci Rep. 2018 Jan 11;8(1):498. evidence of recurrent mural thrombosis with peripheral emboliza­
124. Baek D, Villen J, Shin C, et al. The impact of microRNAs on protein tion culminating in total vascular occlusion. Circulation. 1985
output. Nature. 2008 Sep 4;455(7209):64–71. Apr;71(4):699–708.
125. Pordzik J, Pisarz K, De Rosa S, et al. The potential role of 143. Davies MJ, Thomas AC, Knapman PA, et al. Intramyocardial pla­
platelet-related microRNAs in the development of cardiovascular telet aggregation in patients with unstable angina suffering
events in high-risk populations, including diabetic patients: a sudden ischemic cardiac death. Circulation. 1986 Mar;73
review. Front Endocrinol (Lausanne). 2018;9:74. (3):418–427.
126. Reis SE, Holubkov R, Conrad Smith AJ, et al. Coronary microvascular 144. Frink RJ, Rooney PA, Trowbridge JO, et al. Coronary thrombosis
dysfunction is highly prevalent in women with chest pain in the and platelet/fibrin microemboli in death associated with acute
absence of coronary artery disease: results from the NHLBI WISE myocardial infarction. Br Heart J. 1988 Feb;59(2):196–200.
study. Am Heart J. 2001 May;141(5):735–741. 145. Leach IH, Blundell JW, Rowley JM, et al. Acute ischaemic lesions in
127. Agewall S, Beltrame JF, Reynolds HR, et al. ESC working group death due to ischaemic heart disease. An autopsy study of 333
position paper on myocardial infarction with non-obstructive cor­ cases of out-of-hospital death. Eur Heart J. 1995 Sep;16
onary arteries. Eur Heart J. 2017;38(3):143–153. (9):1181–1185.
•• this is an important article in the field of non-obstructive
146. Dörge H, Neumann T, Behrends M, et al. Perfusion-contraction
coronary disease
mismatch with coronary microvascular obstruction: role of
128. Pasupathy S, Air T, Dreyer RP, et al. Systematic review of patients
inflammation. Am J Physiol Heart Circ Physiol. 2000 Dec;279(6):
presenting with suspected myocardial infarction and nonob­
H2587–92.
structive coronary arteries. Circulation. 2015 Mar 10;131
147. Thielmann M, Dörge H, Martin C, et al. Myocardial dysfunction
(10):861–870.
with coronary microembolization: signal transduction through
129. Montone RA, Niccoli G, Russo M, et al. Clinical, angiographic and
a sequence of nitric oxide, tumor necrosis factor-alpha, and
echocardiographic correlates of epicardial and microvascular
sphingosine. Circ Res. 2002 Apr 19;90(7):807–813.
spasm in patients with myocardial ischaemia and
non-obstructive coronary arteries. Clin Res Cardiol. 2020 Apr;109 148. Li L, Zhao X, Lu Y, et al. Altered expression of pro- and
(4):435–443. anti-inflammatory cytokines is associated with reduced cardiac
130. Pirozzolo G, Seitz A, Athanasiadis A, et al. Microvascular spasm in function in rats following coronary microembolization. Mol Cell
non-ST-segment elevation myocardial infarction without culprit Biochem. 2010 Sep;342(1–2):183–190.
lesion (MINOCA). Clin Res Cardiol. 2020 Feb;109(2):246–254. 149. Arras M, Strasser R, Mohri M, et al. Tumor necrosis factor-alpha is
131. Ong P, Camici PG, Beltrame JF, et al. International standardization expressed by monocytes/macrophages following cardiac microem­
of diagnostic criteria for microvascular angina. Int J Cardiol. 2018 bolization and is antagonized by cyclosporine. Basic Res Cardiol.
Jan 01;250:16–20. 1998 Apr;93(2):97–107.
132. Eggers KM, Hjort M, Baron T, et al. Morbidity and cause-specific 150. Dörge H, Schulz R, Belosjorow S, et al. Coronary Microembolization:
mortality in first-time myocardial infarction with nonobstructive the Role of TNF- α in Contractile Dysfunction. J Mol Cell Cardiol.
coronary arteries. J Intern Med. 2019 04; 285(4): 419–428. 2002 Jan;34(1):51–62.
133. Pelliccia F, Pasceri V, Niccoli G, et al. Predictors of Mortality in 151. Thygesen K, Alpert JS, Jaffe AS, et al. Fourth universal definition
Myocardial Infarction and Nonobstructed Coronary Arteries: of myocardial infarction (2018). Circulation. 2018;138(20):e618–
a Systematic Review and Meta-Regression. Am J Med. 2020 01; e651.
133(1): 73–83.e4. 152. Heusch G. Coronary microvascular obstruction: the new frontier
134. Nordenskjöld AM, Lagerqvist B, Baron T, et al. Reinfarction in in cardioprotection. Basic Res Cardiol. 2019;114(6):45.
patients with myocardial infarction with nonobstructive coronary 153. Sheehan FH, Braunwald E, Canner P, et al. The effect of intravenous
arteries (MINOCA): coronary findings and prognosis. Am J Med. thrombolytic therapy on left ventricular function: a report on
2019 03; 132(3): 335–346. tissue-type plasminogen activator and streptokinase from the
135. Piccirillo F, Carpenito M, Verolino G, et al. Changes of the coronary Thrombolysis in Myocardial Infarction (TIMI Phase I) trial.
arteries and cardiac microvasculature with aging: implications for Circulation. 1987 Apr;75(4):817–829.
translational research and clinical practice. Mech Ageing Dev. 2019 154. Fernández-Jiménez R, García-Prieto J, Sánchez-González J, et al.
12;184:111161. Pathophysiology underlying the bimodal edema phenomenon
362 G. CIMMINO ET AL.

after myocardial ischemia/reperfusion. J Am Coll Cardiol. 2015 Aug 159. Mangiacapra F, Di Gioia G, Pellicano M, et al. Effects of prasugrel
18;66(7):816–828. versus clopidogrel on coronary microvascular function in patients
155. Higginson LA, White F, Heggtveit HA, et al. Determinants of myo­ undergoing elective PCI. J Am Coll Cardiol. 2016;68(2):235–237.
cardial hemorrhage after coronary reperfusion in the anesthetized 160. Barbato E, Sarno G, Berza CT, et al. Impact of alpha- and
dog. Circulation. 1982 Jan;65(1):62–69. beta-adrenergic receptor blockers on fractional flow reserve and
156. Kloner RA, Ganote CE, Jennings RB. The “no-reflow” phenomenon after index of microvascular resistance. J Cardiovasc Transl Res. 2014
temporary coronary occlusion in the dog. J Clin Invest. 1974 Dec;54 Dec;7(9):803–809.
(6):1496–1508. 161. Mangiacapra F, Pellicano M, Di Serafino L, et al. Platelet reac­
157. Di Serafino L, Mangiacapra F, Pyxaras S, et al. Relationship between tivity and coronary microvascular impairment after percuta­
peripheral arterial reactive hyperemia and the index of myocardial neous revascularization in stable patients receiving
resistance in patients undergoing invasive coronary angiography. clopidogrel or prasugrel. Atherosclerosis. 2018 11;278:23–28.
Int J Cardiol. 2021;333:8–13. 162. Bairey Merz CN, Pepine CJ, Walsh MN, et al. Ischemia and no
158. Mangiacapra F, Peace AJ, Di Serafino L, et al. Intracoronary enalaprilat obstructive coronary artery disease (INOCA): developing
to reduce microvascular damage during percutaneous coronary inter­ evidence-based therapies and research agenda for the next
vention (ProMicro) study. J Am Coll Cardiol. 2013;61(6):615–621. decade. Circulation. 2017 Mar 14;135(11):1075–1092.

You might also like